CN111107874A - Combination therapy for cancer - Google Patents

Combination therapy for cancer Download PDF

Info

Publication number
CN111107874A
CN111107874A CN201880060035.0A CN201880060035A CN111107874A CN 111107874 A CN111107874 A CN 111107874A CN 201880060035 A CN201880060035 A CN 201880060035A CN 111107874 A CN111107874 A CN 111107874A
Authority
CN
China
Prior art keywords
amino acid
acid sequence
imid
antigen binding
binding protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201880060035.0A
Other languages
Chinese (zh)
Inventor
S.坎德卡尔
P.梅斯
J.奥帕林斯卡
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline Intellectual Property Development Ltd
Original Assignee
GlaxoSmithKline Intellectual Property Development Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by GlaxoSmithKline Intellectual Property Development Ltd filed Critical GlaxoSmithKline Intellectual Property Development Ltd
Publication of CN111107874A publication Critical patent/CN111107874A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Oncology (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Disclosed herein are methods of treating cancer, such as multiple myeloma, involving a combination of an anti-BCMA antigen binding protein (e.g., an anti-BCMA antibody) and an immunomodulatory drug (e.g., pomalidomide or lenalidomide). The combination may also comprise an anti-inflammatory compound (e.g., dexamethasone).

Description

Combination therapy for cancer
Sequence listing
This application contains a sequence listing that has been electronically filed in ASCII format and is incorporated by reference herein in its entirety. The ASCII copy was created in 2018 on 10/9 under the name PU66429_ WO _ sl.
Technical Field
The present invention relates to methods of treating cancer in a subject. In particular, the invention relates to a combination of an anti-BCMA antigen binding protein and an immunomodulatory imide drug (IMiD) for use in the treatment of cancer. The combination may further comprise an anti-inflammatory compound, such as dexamethasone.
Background
Multiple Myeloma (MM) is an incurable malignancy, accounting for 1% of all cancers and 10% of all hematologic malignancies. A variety of drugs and combination therapies have been evaluated and found to be effective in treating multiple myeloma (National Comprehensive Cancer Network, 2016; Moreau, San Miguel et al, 2017). However, most, if not all, patients inevitably relapse (Richardson, Barlogie et al, 2003; Richardson, Barlogie et al, 2006; Jagannath, Barlogie et al, 2008).
Combinations of three and four drugs have emerged for previously treated MM patients, but these regimens may be limited by toxic effects (National Comprehensive Cancer Network, 2016). There is a need for drugs with new mechanisms of action that can be combined with existing therapies without increasing severe toxicity. Therefore, there is an urgent need to develop a combination of therapies whose mechanisms of action do not overlap, and in which case cross-resistance to previous therapies can be minimized.
Disclosure of Invention
The present disclosure relates to methods of treating cancer in a subject, e.g., a human. In particular, the invention relates to combinations of anti-BCMA antigen binding proteins, such as antibodies, and immunomodulatory imide drugs (imids), for use in the treatment of cancer. The combination may further comprise an anti-inflammatory compound such as dexamethasone. In one embodiment, the cancer is selected from multiple myeloma, chronic lymphocytic leukemia, and non-hodgkin's lymphoma.
Provided herein are methods of treating cancer in a subject in need thereof comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD. In one embodiment, the combination further comprises an anti-inflammatory compound.
Also provided herein are methods of treating cancer in a subject in need thereof comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD, wherein the antibody comprises CDRH1 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID No. 1; CDRH2 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO. 2; 3 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID No. 3; CDRL1 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO. 4; CDRL2 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO. 5; and a CDRL3 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
Also provided herein are methods of treating cancer in a subject in need thereof, comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD, wherein the anti-BCMA antigen binding protein is an antibody comprising a VH comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID No. 7; and a VL comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO. 8.
Provided herein are methods of treating cancer in a subject in need thereof comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, an IMiD, and an anti-inflammatory compound, wherein the anti-inflammatory compound is dexamethasone.
Also provided herein are methods of treating cancer in a subject in need thereof comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD, wherein the IMiD is a thalidomide analog. In one embodiment, the thalidomide analog is lenalidomide or pomalidomide.
Also provided herein are methods of treating cancer in a subject in need thereof, comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD, wherein the anti-BCMA antigen binding protein is an immunoconjugate comprising an antibody conjugated to a cytotoxin. In one embodiment, the cytotoxin is MMAE or MMAF.
Provided herein are methods of treating cancer, wherein 1.9mg/kg, 2.5mg/kg, or 3.4mg/kg of anti-BCMA antigen binding protein is administered on day 1 of a 28-day cycle.
Also provided herein are methods of treating cancer, wherein the IMiD is pomalidomide and wherein 4mg of pomalidomide is administered on days 1 to 21 of a 28-day cycle.
Also provided herein are methods of treating cancer, wherein the IMiD is lenalidomide and wherein 10mg or 25mg of lenalidomide is administered on days 1-21 of a 28-day cycle.
Also provided are methods of treating cancer, wherein the anti-inflammatory compound is dexamethasone and wherein 20mg or 40mg of dexamethasone is administered on days 1-4, 9-12, and 17-20 of a 28-day cycle or on days 1, 8,15, and 22 of a 28-day cycle.
Provided herein are combinations for treating cancer, wherein the combinations comprise an anti-BCMA antigen binding protein, IMiD, and optionally, an anti-inflammatory compound.
Also provided is the use of a combination comprising an anti-BCMA antigen binding protein, an IMiD, and optionally, an anti-inflammatory compound, in the manufacture of a medicament for the treatment of cancer.
Provided herein are kits for treating cancer comprising:
(i) anti-BCMA antigen binding protein;
(ii) instructions for treating cancer when combined with an IMiD and optionally an anti-inflammatory compound.
Also provided are methods of treating cancer in a human in need thereof comprising administering an anti-BCMA antibody drug conjugate, a thalidomide analog, and optionally, an anti-inflammatory compound.
Detailed Description
The present disclosure relates to methods of treating cancer in a subject. In particular, the invention relates to a combination of an anti-BCMA antigen binding protein and IMiD for use in the treatment of cancer. The combination may further comprise an anti-inflammatory compound such as dexamethasone. Without being bound by theory, it is believed that the novel combinations described herein result in reduced toxicity due to non-overlapping mechanisms of action.
Combinations and pharmaceutical compositions
The term "combination" as used herein refers to at least two therapeutic agents. As used herein, the term "therapeutic agent" is understood to refer to a substance that produces a desired effect in a tissue, system, animal, mammal, human, or other subject. In one embodiment, the combination is an anti-BCMA antigen binding protein, suitably an anti-BCMA antibody, and at least one further therapeutic agent. In one embodiment, the combination is an anti-BCMA antigen binding protein and IMiD. In another embodiment, the combination is an anti-BCMA antigen binding protein, an IMiD, and an anti-inflammatory compound. The combinations described herein are effective in treating cancer.
In one embodiment, the combination may comprise other therapeutic agents, for example, other cancer therapeutic agents. In embodiments the other cancer therapeutic is a proteasome inhibitor such as bortezomib, carfilzomib, esxazomib, or oprozomib.
Administration of the combination of the invention may be advantageous over the administration of the individual therapeutic agents, because the combination may provide one or more of the following improved properties compared to the administration of the individual therapeutic agents alone: i) greater anticancer effect than the most effective single drug; ii) synergistic or highly synergistic anti-cancer activity; iii) a dosing regimen that provides enhanced anti-cancer activity with reduced side effects; iv) reduction of toxic effects; v) an increase in the therapeutic window, or vi) an increase in the bioavailability of one or both therapeutic agents.
The combination described herein may be in the form of a pharmaceutical composition. A "pharmaceutical composition" comprises a combination as described herein and one or more pharmaceutically acceptable carriers, diluents, or excipients. The carrier, diluent or excipient must be acceptable in the sense of being compatible with the other ingredients of the formulation, capable of pharmaceutical formulation and not deleterious to the recipient thereof.
In one embodiment, each therapeutic agent in the combination is formulated individually as its own pharmaceutical composition, and each pharmaceutical composition is administered to treat cancer. In this embodiment, each pharmaceutical composition may have the same or different carrier, diluent or excipient. For example, in one embodiment, the first pharmaceutical composition comprises an anti-BCMA antigen binding protein, the second pharmaceutical composition comprises IMiD, and the first and second pharmaceutical compositions are administered to treat cancer. In another embodiment, the first pharmaceutical composition comprises an anti-BCMA antigen binding protein, the second pharmaceutical composition comprises an IMiD, the third pharmaceutical composition comprises an anti-inflammatory compound, and the first, second, and third pharmaceutical compositions are administered separately to treat cancer.
In one embodiment, each therapeutic agent in the combination is formulated together as a single pharmaceutical composition and administered to treat cancer. For example, in one embodiment, a single pharmaceutical composition comprises both an anti-BCMA antigen binding protein and an IMiD and is administered as a single pharmaceutical composition to treat cancer. In another embodiment, a single pharmaceutical composition comprises an anti-BCMA antigen binding protein, an IMiD, and an anti-inflammatory compound, and is administered as a single pharmaceutical composition to treat cancer.
It will be understood that references herein to IMiD and anti-inflammatory compounds refer to IMiD and anti-inflammatory compounds as free bases or salts, such as pharmaceutically acceptable salts. Pharmaceutically acceptable salts include acid addition salts. For an overview of suitable salts, see Berge et al, j.pharm.sci., 66:1-19 (1977).
The present invention includes within its scope all possible stoichiometric and non-stoichiometric forms of IMiD and salts of anti-inflammatory compounds.
It will be appreciated that many organic compounds may form complexes with the solvents in which they react or from which they precipitate or crystallize. These complexes are known as "solvates". For example, complexes with water are referred to as "hydrates". Solvents with high boiling points and/or solvents with a high tendency to form hydrogen bonds, such as water, ethanol, isopropanol, and N-methylpyrrolidone, may be used to form the solvate. Methods of identifying solvation include, but are not limited to, NMR and microanalysis. Solvates of IMiD and anti-inflammatory compounds are within the scope of the invention. As used herein, the term solvate includes solvates of the free base IMiD and the anti-inflammatory compound and any salts thereof.
Certain IMiD and anti-inflammatory compounds of the invention may contain chiral atoms and thus may exist in one or more stereoisomeric forms. The present invention includes all stereoisomers, including optical isomers, of the IMiD and anti-inflammatory compounds of the invention, whether as individual stereoisomers or mixtures thereof, including racemates and mixtures. Any stereoisomer can comprise less than 10 wt.%, e.g., less than 5 wt.%, or less than 0.5 wt.% of any other stereoisomer. For example, any optical isomer may comprise less than 10 wt%, such as less than 5 wt%, or less than 0.5 wt% of its enantiomer.
Certain imids and anti-inflammatory compounds of the invention may exist in tautomeric forms. It is to be understood that the invention encompasses all tautomers of the IMiD and anti-inflammatory compounds of the invention, whether as individual tautomers or as mixtures thereof.
The IMiD and anti-inflammatory compounds of the present invention may be in crystalline or amorphous form. Furthermore, certain crystalline forms of the IMiD and anti-inflammatory compounds of the present invention may exist as polymorphs, all of which are intended to be included within the scope of the present invention. Of particular interest are the most thermodynamically stable polymorphic forms of one or more of the IMiD and anti-inflammatory compounds of the present invention.
The polymorphic forms of the IMiD and anti-inflammatory compounds of the present invention may be characterized and distinguished using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD), infrared spectroscopy (IR), raman spectroscopy, Differential Scanning Calorimetry (DSC), thermogravimetric analysis (TGA), and solid state nuclear magnetic resonance (ssNMR).
The invention also includes all suitable isotopic variations of the IMiD and the anti-inflammatory compound or a pharmaceutically acceptable salt thereof. Isotopic variations of IMiD and anti-inflammatory compounds or pharmaceutically acceptable salts thereof are defined as wherein at least one atom is replaced by a moiety having the same atomic number but an atomic mass different from the atomic mass usually found in natureVariants of the subsubstituted atoms. Examples of isotopes that can be incorporated into imids and anti-inflammatory compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example each2H、3H、13C、14C、15N、17O、18O、18F and36and (4) Cl. Certain isotopic variations of the IMiD and anti-inflammatory compounds or salts or solvates thereof, e.g. wherein a radioactive isotope such as3H or14C, useful in drug and/or substrate tissue distribution studies. Tritium (i.e., tritium) is particularly preferred due to its ease of preparation and detectability3H) And carbon-14 (i.e.14C) An isotope. In addition, with isotopes such as deuterium2H substitution may provide certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and thus may be preferred in certain circumstances. Isotopic variations of IMiD or its pharmaceutical salts can generally be prepared by conventional methods.
From the foregoing, it will be appreciated that solvates, hydrates, isomers and polymorphs of IMiD and anti-inflammatory compounds, and salts and solvates thereof, are included within the scope of the present invention.
One skilled in the art will appreciate that certain derivatives of IMiD and anti-inflammatory compounds, although not necessarily pharmacologically active, may be administered and then metabolized in vivo to form an IMiD and anti-inflammatory compound that is pharmacologically active. These derivatives are referred to herein as "prodrugs". Thus, the IMiD and anti-inflammatory compounds described herein may exist in prodrug form. Examples of suitable derivatives are described in Drugs of Today, vol 19, 9, 1983, pp 499-538 and Topics in chemistry, chapter 31, pp 306-316 and H.Bundgaard "Design of Drugs", Elsevier, 1985, chapter 1.
anti-BCMA antigen binding proteins
The anti-BCMA antigen binding proteins in the combinations described herein are useful for treating or preventing cancer. Any of the anti-BCMA antigen binding proteins disclosed herein can be used in combination with IMiD or in combination with IMiD and an anti-inflammatory compound for the treatment of cancer. The anti-BCMA antigen binding proteins described herein can bind to human BCMA, including, for example, human BCMA comprising the amino acid sequence of GenBank accession No. Q02223.2 or a gene encoding human BCMA having at least 90% homology or at least 90% identity thereto.
The term "antigen binding protein" as used herein refers to antibodies, antibody fragments and other protein constructs capable of binding to human BCMA. The antigen binding protein of the present invention may comprise the heavy chain variable region and the light chain variable region of the present invention, which may be formatted as the structure of a natural antibody or a functional fragment thereof or an equivalent thereof. Thus, an antigen binding protein of the invention may comprise a VH region of the invention formatted as a full length antibody, (Fab')2 fragment, Fab fragment, or equivalent thereof (e.g., scFV, diabody, triabody or tetrabody, Tandab, etc.) when paired with a suitable light chain. The antibody may be IgG1, IgG2, IgG3, or IgG 4; or IgM; IgA, IgE or IgD or modified variants thereof. The constant domains of the antibody heavy chains may be selected accordingly. The light chain constant domain may be a kappa or lambda constant domain. Furthermore, antigen binding proteins may comprise all classes of modifications, e.g., IgG dimers, Fc mutants that no longer bind Fc receptors or mediate C1q binding. The antigen binding protein may also be a chimeric antibody of the type described in WO86/01533 which comprises an antigen binding region and a non-immunoglobulin region.
In another aspect, the antigen binding protein is selected from the group consisting of dAb, Fab, Fab ', F (ab')2Fv, diabody, triabody, tetrabody, minibody and minibody. In one aspect of the invention, the antigen binding protein is a humanized or chimeric antibody, and in another aspect the antibody is humanized. In one aspect the antibody is a monoclonal antibody.
Chimeric Antigen Receptors (CARs) have been developed as artificial T cell receptors to generate new specificities in T cells without the need for binding to MHC antigen peptide complexes. These synthetic receptors contain a target binding domain in a single fusion molecule that is bound to one or more signaling domains by a flexible linker. The target binding domain is used to target T cells to specific targets on the surface of pathological cells, and the signaling domain contains the molecular machinery of T cell activation and proliferation. A flexible linker that crosses the T cell membrane (i.e., forms a transmembrane domain) allows for cell membrane display of the target binding domain of the CAR. CARs have successfully allowed T-cell redirection against antigens expressed at the surface of tumor cells from various malignancies, including lymphomas and solid tumors (Jena et al (2010) Blood,116(7): 1035-44).
To date, the development of CARs has encompassed three generations. The first generation CARs comprise a target binding domain linked to a signaling domain derived from CD3 ζ or a cytoplasmic region of the Fc receptor γ chain. First generation CARs were shown to successfully redirect T cells to selected targets, however, they failed to provide prolonged expansion and anti-tumor activity in vivo. Second and third generation CARs have focused on enhancing modified T cell survival and increasing proliferation by including co-stimulatory molecules such as CD28, OX-40(CD134), and 4-1BB (CD 137).
CAR-bearing T cells can be used to eliminate pathological cells in the disease background. One clinical goal is to transform patient cells with recombinant DNA containing the CAR expression construct via a vector (e.g., a lentiviral vector) after apheresis (aphaeresis) and T cell isolation. After T cells are expanded, they are reintroduced into the patient to target and kill the pathological target cells.
In one aspect of the invention, the anti-BCMA antigen binding protein is a chimeric antigen receptor. In another aspect, the CAR comprises a binding domain, a transmembrane domain, and an intracellular effector domain.
For example, the transmembrane domain may be a CD, such as the transmembrane domain of a CD4, CD8, CD3, or CD28 protein, a subunit of a T cell receptor such as α, β, gamma, or delta, a subunit of an IL-2 receptor (α chain), a subunit of a low affinity nerve growth factor receptor (LNGFR or p75) (β chain or gamma chain), or a subunit chain of an Fc receptor.
In another aspect, the transmembrane domain comprises a transmembrane domain of CD4 or CD8 (e.g., a CD8 α chain, as described in NCBI reference sequence: NP _0011 45.1, incorporated herein by reference).
The intracellular effector domain or "signaling domain" is responsible for intracellular signaling upon binding of the target binding domain to the target. The intracellular effector domain is responsible for activating at least one normal effector function of the CAR-expressing immune cell. For example, the effector function of a T cell may be cytolytic activity or helper activity, including secretion of cytokines. Preferred examples of effector domains for CAR scaffolds may be the cytoplasmic sequences of the native T cell receptor and co-receptors that act synergistically to initiate signal transduction upon antigen binding, as well as any derivative or variant of these sequences, and any synthetic sequence with the same functional capabilities.
Effector domains can be divided into two classes, those that initiate antigen-dependent primary activation, and those that function in an antigen-independent manner to provide secondary or costimulatory signals primary activating effector domains can comprise signaling motifs known as immunoreceptor tyrosine-based activation motifs (ITAMs) ITAMs are well-defined signaling motifs that are typically present at the intracytoplasmic tails of a variety of receptors and serve as binding sites for tyrosine kinases of the syk/zap70 class examples of ITAMs for use in the present invention can include, as non-limiting examples, those derived from CD3 ζ, FcR γ, FcR β, FcR ε, CD3 γ, CD3 δ, CD3 ε, CD5, CD22, CD79a, CD79b, and CD66 d.
In one aspect of the invention, the intracellular signaling domain is a CD3 zeta effector domain. The effector domain may also provide a secondary or co-stimulatory signal. In addition, T cells contain co-stimulatory molecules that bind to cognate co-stimulatory ligands on antigen presenting cells to enhance T cell responses, e.g., by increasing proliferation activation, differentiation, etc. Thus, in one aspect, the intracellular effector domain additionally comprises a co-stimulatory domain. In another aspect, the co-stimulatory domain comprises an intracellular domain of a co-stimulatory molecule selected from the group consisting of CD28, CD27, 4-1BB (CD137), OX40(CD134), ICOS (CD278), CD30, CD40, PD-1(CD279), CD2, CD7, NKG2C (CD94), B7-H3(CD276), or any combination thereof. In yet another aspect, the co-stimulatory domain comprises an endodomain of a co-stimulatory molecule selected from the group consisting of CD28, CD27, 4-1BB, OX40, ICOS, or any combination thereof.
Exemplary anti-BCMA antigen binding proteins and methods of making the same are disclosed in international publication No. WO2012/163805, which is incorporated by reference herein in its entirety. Other exemplary anti-BCMA antigen binding proteins include those described in WO2016/014789, WO2016/090320, WO2016/090327, WO2016/020332, WO2016/079177, WO2014/122143, WO2014/122144, WO2017/021450, WO2016/014565, WO2014/068079, WO2015/166649, WO2015/158671, WO 20152014/052536, WO2014/140248, WO2013/072415, WO2013/072406, WO2014/089335, US2017/165373, WO2013/154760, and WO2017/051068, each of which is incorporated herein by reference in its entirety.
In one embodiment, the anti-BCMA antigen binding protein has enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) effector function. As used herein, the term "effector function" refers to one or more of antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) mediated response, Fc-mediated phagocytosis, and antibody recycling via FcRn receptors. For IgG antibodies, effector functions (including ADCC and ADCP) are mediated by the interaction of the heavy chain constant region with a class of Fc γ receptors present on the surface of immune cells. In humans, these include Fc γ RI (CD64), Fc γ RII (CD32) and Fc γ RIII (CD 16). The interaction between the antigen binding protein bound to the antigen and the formation of the Fc/fcgamma complex results in a range of effects including cytotoxicity, immune cell activation, phagocytosis, and release of inflammatory cytokines.
In another embodiment, the anti-BCMA antigen binding protein described herein inhibits the binding of BAFF and/or APRIL to the BCMA receptor. In another embodiment, the anti-BCMA antigen binding protein described herein is capable of binding to Fc γ RIIIA or is capable of Fc γ RIIIA mediated effector function.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a heavy chain variable region CDR1 ("CDRH 1") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 1. In one embodiment, the heavy chain variable region CDR1 ("CDRH 1") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID NO: 1.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a heavy chain variable region CDR2 ("CDRH 2") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 2. In one embodiment, the heavy chain variable region CDR2 ("CDRH 2") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID NO: 2.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a heavy chain variable region CDR3 ("CDRH 3") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 3. In one embodiment, the heavy chain variable region CDR3 ("CDRH 3") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID NO: 3.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a light chain variable region CDR1 ("CDRL 1") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 4. In one embodiment, the light chain variable region CDL1 ("CDR 1") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID NO: 4.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a light chain variable region CDR2 ("CDRL 2") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 5. In one embodiment, the light chain variable region CDL2 ("CDR 2") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID NO: 5.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a light chain variable region CDR3 ("CDRL 3") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 6. In one embodiment, the light chain variable region CDL3 ("CDR 3") comprises an amino acid sequence having one amino acid change (variant) from the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a CDRH1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 1; a CDRH2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 2; 3, a CDRH3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO; a CDRL1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in seq id No. 4; (ii) a CDRL2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 5; and/or a CDRL3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a heavy chain variable region ("VH") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 7.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a light chain variable region ("VL") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 8.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a VH comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 7; and a VL comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 8.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a heavy chain region ("HC") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 9.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising a light chain region ("LC") comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 10.
In one embodiment, the anti-BCMA antigen binding protein is an antibody comprising an HC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 9; and an LC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 10.
In one embodiment, the anti-BCMA antigen binding protein is an immunoconjugate comprising an antigen binding protein according to the invention as described herein, including, but not limited to, an antibody conjugated to one or more cytotoxic agents, such as a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., a protein toxin, an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or a fragment thereof), or a radioisotope (i.e., a radioconjugate). In another embodiment, the anti-BCMA antigen binding protein is conjugated to a toxin such as an auristatin, e.g., monomethyl auristatin e (mmae) or monomethyl auristatin f (mmaf).
In one embodiment, the anti-BCMA antigen binding protein is an immunoconjugate having the general structure:
ABP- ((linker)n-Ctx)m
Wherein
ABP is an antigen binding protein
The linker being absent or any cleavable or non-cleavable linker
Ctx is any cytotoxic agent described herein
n is 0, 1, 2 or 3, and
m is 1, 2, 3, 4, 5,6, 7,8, 9 or 10.
Exemplary linkers include 6-Maleimidocaproyl (MC), Maleimidopropanoyl (MP), valine-citrulline (val-cit), alanine-phenylalanine (ala-phe), p-aminobenzyloxycarbonyl (PAB), N-succinimidyl 4- (2-pyridylthio) pentanoate (SPP), N-succinimidyl 4- (N-maleimidomethyl) cyclohexane-1 carboxylate (SMCC), and N-succinimidyl (4-iodo-acetyl) aminobenzoate (SIAB).
In one embodiment, the anti-BCMA antigen binding protein is an immunoconjugate comprising a monoclonal antibody linked to MMAE or MMAF. In another embodiment, the anti-BCMA antigen binding protein is an immunoconjugate comprising a monoclonal antibody linked to MMAE or MMAF through an MC linker, as shown in the structure:
Figure BDA0002412797080000121
an appropriate therapeutically effective dose of anti-BCMA antigen binding protein will be readily determined by one skilled in the art. As used herein, the term "effective dose" refers to a dose of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for example, by a researcher or clinician. Furthermore, the term "therapeutically effective dose" refers to any dose that results in an improved treatment, cure, prevention, or amelioration, or reduction in the rate of progression of a disease or disorder as compared to a corresponding subject that does not receive the dose. The term also includes within its scope dosages effective to enhance normal physiological function.
Suitable doses of the anti-BCMA antigen binding proteins described herein may be calculated for the patient based on the body weight thereof, e.g. suitable doses may range from about 0.1 to about 20mg/kg, e.g. from about 1 to about 20mg/kg, e.g. from about 10 to about 20mg/kg or e.g. from about 1 to about 15mg/kg, e.g. from about 10 to about 15 mg/kg.
In one embodiment, the therapeutically effective dose of anti-BCMA antigen binding protein ranges from about 0.03mg/kg to about 4.6 mg/kg. In another embodiment, the therapeutically effective dose of the anti-BCMA antigen binding protein is 0.03mg/kg, 0.06mg/kg, 0.12mg/kg, 0.24mg/kg, 0.48mg/kg, 0.96mg/kg, 1.92mg/kg, 3.4mg/kg, or 4.6 mg/kg. In another embodiment, the therapeutically effective dose of anti-BCMA antigen binding protein is 1.9mg/kg, 2.5mg/kg, or 3.4 mg/kg.
Immunomodulatory imine drugs (IMiD)
The term "immunomodulatory imine drug (IMiD)" as used herein refers to a class of drugs that contain an imide group. Without being bound by theory, it is believed that IMiD may be useful in the treatment of cancer due to immunomodulatory, anti-angiogenic and anti-tumor properties. The class of ImiD drugs includes, but is not limited to, thalidomide and its analogs. As used herein, the term "analog" is a compound that has a similar structure to another compound, but differs therefrom with respect to certain constituents, e.g., an analog may differ in one or more atoms, functional groups, or substructures that are replaced with other atoms, groups, or substructures. Such structural differences can be at least theoretically imagined by the person skilled in the art from another compound.
Various imids are known to those skilled in the art, including, for example, thalidomide, lenalidomide, pomalidomide, apremilast, and the like.
In one embodiment, the IMiD comprises thalidomide or an analog thereof. The registered trade name of thalidomide is
Figure BDA0002412797080000132
(Celgene Corp) and has the following chemical structure:
Figure BDA0002412797080000131
thalidomide and its analogs, and methods for their preparation, are known to those skilled in the art, for example, U.S. patent nos. 6,045,501; 7,230,012, respectively; 7,435,745, the disclosure of which is incorporated herein in its entirety.
In another embodiment, the IMiD comprises pomalidomide or an analogue thereof. Registered trade name of pomalidomide
Figure BDA0002412797080000144
(Celgene Corp) and has the following chemical structure:
Figure BDA0002412797080000141
pomalidomide and its analogs, and methods for their preparation, are known to those skilled in the art, for example, U.S. Pat. nos. 5,635,517; 6,316,471; 6,476,052, respectively; 8,158,653, respectively; 8,198,262, respectively; 8,673,939, respectively; 8,735,428, respectively; and 8,828,427, the disclosures of which are incorporated herein in their entirety.
In another embodiment, the IMiD comprises lenalidomide or an analog thereof. The registered trade name of lenalidomide is
Figure BDA0002412797080000145
(Celgene Corp) and has the following chemical structure:
Figure BDA0002412797080000142
lenalidomide and analogs thereof, and methods of making the same, are known to those skilled in the art, for example, U.S. Pat. nos. 5,635,517; 6,555,554, respectively; 7,119,106, respectively; 7,465,800, respectively; 7,855,217, respectively; 8,288,415, respectively; and 8,530,498, the disclosures of which are incorporated herein in their entirety.
In another embodiment, the IMiD is apremilast or an analog thereof. Registered trade name of Apremilast
Figure BDA0002412797080000146
(Celgene Corp) and has the following chemical structure:
Figure BDA0002412797080000143
apremilast and its analogs, and methods of making the same, are known to those skilled in the art, for example, U.S. Pat. nos. 6,020,358; 7,427,638, respectively; 7,893,101, the disclosure of which is incorporated herein in its entirety.
Suitable therapeutically effective dosages of IMiD are readily determined by those skilled in the art. Suitable dosages of imids described herein may be calculated for a patient based on their body weight. A therapeutically effective dose is typically about 1 to 2000mg, 5 to 2000mg, 10 to 2000mg and suitably about 30 to 1500 mg. Other ranges may be used, including, for example, 50-500mg, 50-300mg, 50-100mg, 100-200mg, 5-100mg, 5-50 mg. Therapeutically effective doses for acute or chronic human treatment range from 0.01 to 250mg/kg body weight, suitably 0.1-5mg/kg body weight, suitably 0.1-10mg/kg body weight, suitably 2-100mg/kg body weight, or suitably 5-60mg/kg body weight, which may be administered, for example, in 1 to 4 daily doses, depending on the route of administration and the condition of the subject.
In one embodiment, the IMiD is thalidomide and the therapeutically effective dose ranges from about 25mg to about 300 mg. In another embodiment, the IMiD is thalidomide and the therapeutically effective dose is 50mg, 100mg, 150mg, or 200 mg. In another embodiment, the IMiD is thalidomide and the therapeutically effective dose is 200 mg.
In one embodiment, the IMiD is pomalidomide and the therapeutically effective dose is in the range from about 0.5mg to about 5mg. In another embodiment, the IMiD is pomalidomide and the therapeutically effective dose is selected from 1mg, 2mg, 3mg, or 4mg. In another embodiment, the IMiD is pomalidomide and the therapeutically effective dose is 4mg.
In one embodiment, the IMiD is lenalidomide and the therapeutically effective dose ranges from about 1mg to about 50 mg. In another embodiment, the IMiD is lenalidomide and the therapeutically effective dose is 2.5mg, 5mg, 10mg, 15mg, 20mg, or 25 mg. In another embodiment, the IMiD is lenalidomide and the therapeutically effective dose is 10mg or 25 mg.
In one embodiment, the IMiD is apremilast and the therapeutically effective dose ranges from about 1mg to about 100 mg. In another embodiment, the IMiD is apremilast and the therapeutically effective dose is 10mg, 20mg, or 30 mg.
Anti-inflammatory compounds
Anti-inflammatory compounds (e.g., dexamethasone) are compounds that reduce inflammation or swelling in various parts of the human body. Anti-inflammatory compounds have been used to reduce swelling (edema) associated with spinal and brain tumors, and to treat ocular inflammation and to treat a variety of cancers, such as leukemia, lymphoma, and multiple myeloma. A variety of anti-inflammatory compounds and methods for their preparation are known to those skilled in the art.
Anti-inflammatory compounds may include steroids and non-steroidal compounds (NSAIDs).
In one embodiment, the anti-inflammatory compound is a steroid. Examples of steroids include, but are not limited to, cortisone, cortisol, corticosterone, hydrocortisone (hydrocortisone), prednisone, prednisolone, dexamethasone, beclomethasone, betamethasone, mometasone furoate, budesonide, triamcinolone acetonide, and fluticasone. In one embodiment, the anti-inflammatory compound is an adrenal corticosteroid selected from dexamethasone, prednisone, prednisolone, methylprednisolone, and methylprednisolone.
In another embodiment, the anti-inflammatory compound is dexamethasone. Dexamethasone has the following chemical structure and is registered under the trade name dexamethasone
Figure BDA0002412797080000162
(Merck&Co.,Inc.):
Figure BDA0002412797080000161
In another embodiment, the anti-inflammatory compound is an NSAID. Examples of NSAIDs useful in the present invention include, but are not limited to, aspirin, acetaminophen, ibuprofen, esculetin, phenidone, quercetin, ketoprofen, nordihydroguaiaretic acid (NDGA), sulindac, indomethacin, NS-398 (a cyclooxygenase-2 inhibitor), cyclooxygenase-1 inhibitor, methylheptylimidazole, furgregaric acid sodium, SKF525AHCL, a thromboxane inhibitor, ketorolac tromethamine, ecasa, salsalate, diflunisal, mefenamic acid, naproxen sodium, frofenonine, meclofenamic acid, phenylbutazone, oxybutyzone, diclofenac, etodolac, fenoprofen, flufenamic acid, flurbiprofen, pirprofen, tolmetin, azapropazone, fenbufen, nabumetone, oxaprozin, piroxicam, a salicylate, and tenoxicam. Preferred NSAIDs are sulindac, sulindac sulfide, indomethacin, NS-398, methylheptylimidazole, sodium furaldehyde, and SKF525 AHCL. Particularly preferred NSAIDs are indomethacin and sulindac.
Suitable therapeutically effective dosages of the anti-inflammatory compounds can be readily determined by those skilled in the art. Suitable dosages of the anti-inflammatory compounds described herein may be calculated for the patient based on their body weight. A therapeutically effective dose is typically about 1 to 2000mg, 5 to 2000mg, 10 to 2000mg and suitably about 30 to 1500 mg. Other ranges may be used, including, for example, 50-500mg, 50-300mg, 50-100mg, 100-200mg, 5-100mg, 5-50 mg. The daily dose for acute or chronic human treatment ranges from 0.01 to 250mg/kg body weight, suitably 0.1-5mg/kg body weight, suitably 0.1-10mg/kg body weight, suitably 2-100mg/kg body weight, or suitably 5-60mg/kg body weight, which may be administered in 1 to 4 daily doses, for example, depending on the route of administration and the condition of the subject.
In one embodiment, the anti-inflammatory compound is dexamethasone and the therapeutically effective dose is about 5mg to about 100 mg. In another embodiment, the anti-inflammatory compound is dexamethasone and the therapeutically effective dose is 20mg or 40 mg.
Method of treatment
Described herein are methods of treating cancer in a subject with a combination described herein. As used herein, the terms "cancer" and "tumor" are used interchangeably and, in either the singular or plural, refer to a cell that has undergone malignant transformation that renders it pathological to a host organism. Primary cancer cells can be readily distinguished from non-cancer cells by well-established techniques, particularly histological examination. The definition of cancer cells as used herein includes not only primary cancer cells, but also any cells derived from a cancer cell progenitor. This includes metastasized cancer cells, as well as in vitro cultures and cell lines derived from cancer cells. When referring to a type of cancer that usually manifests as a solid tumor, a "clinically detectable" tumor is one that is detectable based on tumor mass; for example, by a program such as a Computed Tomography (CT) scan, Magnetic Resonance Imaging (MRI), X-ray, ultrasound, or palpation at the time of physical examination, and/or it may be detectable due to expression of one or more cancer specific antigens in a sample obtainable from the patient. The tumor may be a hematopoietic (or hematologic or blood-related) cancer, such as a cancer derived from blood cells or immune cells, which may be referred to as a "liquid tumor. Specific examples of hematological tumor-based clinical conditions include: leukemias, such as chronic myelogenous leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, and acute lymphocytic leukemia; plasma cell malignancies such as multiple myeloma, MGUS, and waldenstrom's macroglobulinemia; lymphomas such as non-hodgkin lymphoma, hodgkin lymphoma; and the like.
The cancer may be any cancer in which there is an abnormal number of blasts of interest or unwanted cellular proliferation or which is diagnosed as a hematological cancer (including lymphoid and myeloid malignancies). Myeloid malignancies include, but are not limited to: acute myeloid (or myelocytic or promyelocytic) leukemia (undifferentiated or differentiated), acute promyelocytic (or promyelocytic) leukemia, acute myelomonocytic (or myelomonocytic) leukemia, acute monocytic (or monocytic) leukemia, erythrocytic leukemia, and megakaryocytic (or megakaryoblastic) leukemia. These leukemias may be collectively referred to as acute myeloid (or myelocytic) leukemia (AML). Myeloid malignancies also include myeloproliferative disorders (MPD), which include, but are not limited to: chronic myelogenous (or myelogenous) leukemia (CML), chronic myelomonocytic leukemia (CMML), essential thrombocythemia (or thrombocythemia), and polycythemia vera (PCV). Myeloid malignancies also include: myelodysplasia (or myelodysplastic syndrome or MDS), which may be referred to as Refractory Anemia (RA), refractory anemia with excess blasts (RAEB), and refractory anemia with excess blasts in transformation (RAEBT); and Myelofibrosis (MFS) with or without agnogenic myeloid metaplasia.
Hematopoietic cancers also include lymphoid malignancies, which can affect lymph nodes, spleen, bone marrow, peripheral blood, and/or extranodal sites. Lymphoid cancers include B-cell malignancies including, but not limited to, B-cell non-Hodgkin's lymphoma (B-NHL). B-NHL can be inert (or low), moderate (or aggressive), or high (highly aggressive). Indolent B-cell lymphomas include: follicular Lymphoma (FL); small Lymphocytic Lymphoma (SLL); marginal Zone Lymphoma (MZL) comprising nodal MZL, extranodal MZL, spleen MZL, and spleen MZL with villous lymphocytes; lymphoplasmacytic lymphoma (LPL); and mucosa-associated lymphoid tissue (MALT or extranodal marginal zone) lymphomas. Moderate B-NHL includes: mantle Cell Lymphoma (MCL), diffuse large cell lymphoma (DLBCL), follicular large cell (or grade 3 or 3B) lymphoma, and Primary Mediastinal Lymphoma (PML), with or without leukemia. High grade B-NHL includes Burkitt's Lymphoma (BL), Burkitt's like lymphoma, small lytic cell-free lymphoma (SNCCL) and lymphoblastic lymphoma. Other B-NHLs include immunoblastic lymphoma (or immunocytoma), primary effusion lymphoma, HIV-related (or AIDS-related) lymphoma, and post-transplant lymphoproliferative disorder (PTLD) or lymphoma. B cell malignancies also include, but are not limited to: chronic Lymphocytic Leukemia (CLL), prolymphocytic leukemia (PLL), Waldenstrom's Macroglobulinemia (WM), Hairy Cell Leukemia (HCL), Large Granular Lymphocytic (LGL) leukemia, acute lymphocytic (or lymphoblastic) leukemia, and castleman's disease. NHL may also include: t-cell non-Hodgkin's lymphoma (T-NHL) including but not limited to T-cell non-Hodgkin's lymphoma Not Otherwise Specified (NOS), peripheral T-cell lymphoma (PTCL), Anaplastic Large Cell Lymphoma (ALCL), angioimmunoblastic lymphoid disorder (AILD), nasal Natural Killer (NK) cell/T-cell lymphoma, gamma/delta lymphoma, cutaneous T-cell lymphoma, mycosis fungoides and Sezary syndrome (Sezary syndrome).
Hematopoietic cancers also include hodgkin's lymphoma (or disease), which includes classical hodgkin's lymphoma, nodal sclerosing hodgkin's lymphoma, mixed cell hodgkin's lymphoma, Lymphocyte Predominant (LP) hodgkin's lymphoma, nodal LP hodgkin's lymphoma and lymphocyte depleting hodgkin's lymphoma. Hematopoietic cancers also include plasma cell diseases or cancers such as Multiple Myeloma (MM), including stasis-type MM, monoclonal gammopathy of undetermined significance (or unknown), plasmacytoma (bone, extramedullary), lymphoplasmacytoma (LPL), waldenstrom's macroglobulinemia, plasma cell leukemia and primary Amyloidosis (AL). Hematopoietic cancers may also include other cancers in which additional hematopoietic cells are present, including polymorphonuclear leukocytes (or neutrophils), basophils, eosinophils, dendritic cells, platelets, erythrocytes and natural killer cells. Tissues comprising hematopoietic cells, referred to herein as "hematopoietic cell tissues," including bone marrow; peripheral blood; thymus; and peripheral lymphoid tissue such as spleen, lymph nodes, mucosa-associated lymphoid tissue (e.g., intestine-associated lymphoid tissue), tonsils, peyer's patches and appendices, and other mucosa-associated lymphoid tissue, e.g., the bronchial lining.
The term "treatment" and derivatives thereof as used herein includes therapeutic treatment. In reference to a particular condition, treatment means (1) ameliorating the condition or one or more of the biological clinical manifestations of the condition; (2) interfering with (a) one or more points in the biological cascade that causes or contributes to the condition or (b) one or more biological clinical manifestations of the condition; (3) alleviating one or more symptoms, effects or side effects associated with the condition, or one or more symptoms, effects or side effects associated with the condition or treatment thereof; (4) slowing the progression of the condition or one or more biological manifestations of the condition and/or (5) curing the condition or one or more biological manifestations of the condition by eliminating or reducing the one or more biological manifestations of the condition to undetectable levels for a period of time, wherein the period of time is considered remission, without additional treatment during the remission period. Those skilled in the art will understand the duration of remission believed to be directed to a particular disease or condition.
Prophylactic treatment is also contemplated. The skilled person will appreciate that "prevention" is not an absolute term. In medicine, "prevention" is understood to mean prophylactic administration of a drug to significantly reduce the likelihood or severity of, or delay the onset of, a condition or its biological clinical manifestations. Prophylactic treatment is appropriate, for example, when the subject is considered to be at high risk of developing cancer, such as when the subject has a strong family history of cancer or when the subject is exposed to carcinogens.
A "subject" is broadly defined to include any patient in need of treatment, e.g., a patient in need of cancer treatment. The subject may comprise a mammal. In one embodiment, the subject is a human patient. Subjects in need of cancer treatment may include patients from various stages, including new diagnosis, relapse, refractory, progressive disease, remission, and the like. Subjects in need of cancer treatment may also include patients who have undergone stem cell transplantation or who are deemed unsuitable for transplantation.
The subject may be pre-screened to select for treatment with a combination as described herein. In one embodiment, a sample from a subject is tested for BCMA expression prior to treatment with the combination described herein.
The subject may have received at least one prior cancer treatment prior to treatment with the combination of the invention. In one embodiment, the subject has received at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, or at least 7 prior cancer treatments prior to treatment with the combination of the invention.
In another embodiment, the subject has a newly diagnosed cancer and has received 0 prior treatments prior to treatment with the combination of the invention.
The individual therapeutic agents of the combination of the invention and the pharmaceutical composition comprising such therapeutic agents may be administered together or separately. When administered separately, this may occur simultaneously or sequentially in any order (by the same or different routes of administration). Such sequential administration may be close in time or distant in time. The dosages and relative administration times of the therapeutic agent of the present invention, or a pharmaceutically acceptable salt thereof, and the other therapeutically active agent will be selected so as to achieve the desired combined therapeutic effect.
The therapeutic agents of the present invention may be administered by any suitable route. For certain therapeutic agents, suitable routes include oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intrathecal and epidural). It will be appreciated that the preferred route may vary depending on, for example, the condition of the recipient of the combination and the cancer to be treated. It is also understood that each agent administered may be administered by the same or different routes, and that the therapeutic agents may be formulated together or in separate pharmaceutical compositions.
In one embodiment, the one or more therapeutic agents of the combination of the invention are administered intravenously. In another embodiment, the one or more therapeutic agents of the COMBINATION OF THE INVENTION are administered intratumorally. In another embodiment, the one or more therapeutic agents of the combination of the invention are administered orally. In another embodiment, the one or more therapeutic agents of the COMBINATION OF THE INVENTION are administered systemically, e.g. intravenously, and the one or more other therapeutic agents of the COMBINATION OF THE INVENTION are administered intratumorally. In another embodiment, all therapeutic agents of the combination of the invention are administered systemically, e.g., intravenously. In an alternative embodiment, all the therapeutic agents of the combination of the invention are administered intratumorally. In any embodiment, e.g., in this paragraph, the therapeutic agents of the present invention are administered as one or more pharmaceutical compositions.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination described herein.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an IMiD.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, an IMiD and an anti-inflammatory compound.
In one embodiment, the invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises a CDRH1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 1; a CDRH2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 2; 3, a CDRH3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO; a CDRL1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in seq id No. 4; (ii) a CDRL2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 5; and/or a CDRL3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises a VH comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 7; and/or a VL comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 8.
In one embodiment, the invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises an HC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ id No. 9; and/or an LC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 10.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, an IMiD, and an anti-inflammatory compound, wherein the anti-BCMA antibody comprises a CDRH1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO: 1; a CDRH2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 2; 3, a CDRH3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO; (ii) a CDRL1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 4; (ii) a CDRL2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 5; and/or a CDRL3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, an IMiD, and an anti-inflammatory compound, wherein the anti-BCMA antibody comprises an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises a VH comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 7; and/or a VL comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 8.
In one embodiment, the invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, an IMiD, and an anti-inflammatory compound, wherein the anti-BCMA antibody comprises an HC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 9; and/or an LC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 10.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and thalidomide.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, thalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and pomalidomide.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, pomalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and lenalidomide.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, lenalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and apremilast.
In one embodiment, the present invention provides a method of treating cancer in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein, apremilast, and an anti-inflammatory compound.
In one embodiment, the present invention provides a method of treating multiple myeloma in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, thalidomide and dexamethasone. In another embodiment, the invention provides a method of treating multiple myeloma in a subject in need thereof by administering 1.9mg/kg, 2.5mg.kg, or 3.4mg/kg of an anti-BCMA antibody, 200mg of thalidomide, and 20mg or 40mg of dexamethasone.
In one embodiment, the present invention provides a method of treating multiple myeloma in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, pomalidomide and dexamethasone. In another embodiment, the invention provides a method of treating multiple myeloma in a subject in need thereof by administering 1.9mg/kg, 2.5mg.kg, or 3.4mg/kg of an anti-BCMA antibody, 4mg of pomalidomide, and 20mg or 40mg of dexamethasone.
In one embodiment, the present invention provides a method of treating multiple myeloma in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, lenalidomide and dexamethasone. In another embodiment, the invention provides a method of treating multiple myeloma in a subject in need thereof by administering 1.9mg/kg, 2.5mg.kg, or 3.4mg/kg of an anti-BCMA antibody, 10mg or 25mg lenalidomide, and 20mg or 40mg dexamethasone.
In one embodiment, the present invention provides a method of treating multiple myeloma in a subject in need thereof by administering a therapeutically effective dose of a combination comprising an anti-BCMA antibody, apremilast, and dexamethasone.
In one embodiment, the present invention provides a combination as described herein for use in therapy.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and an IMiD.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, an IMiD and an anti-inflammatory compound.
In one embodiment, the invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises a CDRH1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO: 1; a CDRH2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 2; 3, a CDRH3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO; (ii) a CDRL1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 4; (ii) a CDRL2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 5; and/or a CDRL3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein said combination comprises an anti-BCMA antibody and an IMiD, wherein said anti-BCMA antibody comprises a VH comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 7; and/or a VL comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 8.
In one embodiment, the invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antibody and an IMiD, wherein the anti-BCMA antibody comprises a HC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO: 9; and/or an LC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 10.
In one embodiment, the invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antibody, an IMiD and an anti-inflammatory compound, wherein the anti-BCMA antibody comprises a CDRH1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 1; a CDRH2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 2; 3, a CDRH3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO; (ii) a CDRL1 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 4; (ii) a CDRL2 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 5; and/or a CDRL3 comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 6.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein said combination comprises an anti-BCMA antibody, an IMiD and an anti-inflammatory compound, wherein said anti-BCMA antibody comprises a VH comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 7; and/or a VL comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 8.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antibody, an IMiD and an anti-inflammatory compound, wherein the anti-BCMA antibody comprises an HC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID No. 9; and/or an LC comprising an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence set forth in SEQ ID NO 10.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and thalidomide.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, thalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and pomalidomide.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, pomalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and lenalidomide.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, lenalidomide and an anti-inflammatory compound.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and apremilast.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, apremilast and an anti-inflammatory compound.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises an anti-BCMA antibody, thalidomide and dexamethasone. In another embodiment, the invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises 1.9mg/kg, 2.5mg/kg or 3.4mg/kg of an anti-BCMA antibody; 200mg of thalidomide; and 20mg or 40mg dexamethasone.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises an anti-BCMA antibody, pomalidomide and dexamethasone. In another embodiment, the invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises 1.9mg/kg, 2.5mg/kg or 3.4mg.kg of an anti-BCMA antibody; 4mg pomalidomide; and 20mg or 40mg dexamethasone.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises an anti-BCMA antibody, lenalidomide and dexamethasone. In another embodiment, the invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises 1.9mg/kg, 2.5mg/kg or 3.4mg.kg of an anti-BCMA antibody; 10mg or 25mg lenalidomide; and 20mg or 40mg dexamethasone.
In one embodiment, the present invention provides a combination as described herein for use in the treatment of multiple myeloma, wherein said combination comprises an anti-BCMA antibody, apremilast and dexamethasone.
In one embodiment, there is provided the use of a combination in the manufacture of a medicament for the treatment of cancer. In another embodiment, there is provided the use of a combination for the manufacture of a medicament for the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein and an IMiD. In another embodiment, there is provided the use of a combination for the manufacture of a medicament for the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, an IMiD and an anti-inflammatory compound.
Treatment regimens
Suitable treatment regimens for anti-BCMA antigen binding proteins, imids and anti-inflammatory compounds are readily determined by those skilled in the art.
In an exemplary treatment regimen, one dose of anti-BCMA antigen binding protein is administered every 3 weeks (21 day cycle) for up to 16 cycles. In another exemplary treatment regimen, one dose of anti-BCMA antigen binding protein is administered once a week for three consecutive weeks, followed by a 1-week rest period (28-day period) for up to 16 cycles. In another exemplary treatment regimen, one dose of anti-BCMA antigen binding protein is administered on the first day of a 28-day cycle.
In an exemplary embodiment, the IMiD is thalidomide and the treatment regimen comprises administering a single dose daily for 28 days for at least one 28-day cycle. In another embodiment, the IMiD is thalidomide and the treatment regimen comprises administration of 200mg on days 1-28 of a 28 day cycle.
In an exemplary embodiment, the IMiD is lenalidomide and the treatment regimen comprises administering a single dose on each of days 1-21 of a 28-day cycle. In another exemplary embodiment, the IMiD is lenalidomide and the treatment regimen comprises administering 25mg on each of days 1-21 of a 28-day cycle. In another exemplary embodiment, the IMiD is lenalidomide and the treatment regimen comprises administering 10mg on each of days 1-21 of a 28-day cycle.
In an exemplary embodiment, the IMiD is pomalidomide and the treatment regimen comprises administering a single dose on each of days 1 to 21 of a 28-day cycle. In another exemplary embodiment, the IMiD is pomalidomide and the treatment regimen comprises administering 4mg on each of days 1 to 21 of a 28-day cycle.
In an exemplary embodiment, the anti-inflammatory compound is dexamethasone and the treatment regimen includes administering a dose of dexamethasone on days 1-4, 9-12, and 17-20 of the 28-day cycle. In another exemplary embodiment, the anti-inflammatory compound is dexamethasone and the treatment regimen comprises administering a dose of dexamethasone on days 1, 8,15, and 22 of the 28-day cycle. In another embodiment, the anti-inflammatory compound is dexamethasone and the treatment regimen comprises administering dexamethasone on days 1, 2, 4, 5,8, 9, 11, and 12 of the 21-day cycle.
In an exemplary treatment regimen, the treatment regimen comprises administering 1.9mg/kg, 2.5mg/kg, or 3.4mg/kg of an anti-BCMA antigen binding protein on day 1 of a 28-day cycle; 4mg pomalidomide on days 1 to 21 of a 28 day cycle; and optionally, administering 20mg or 40mg of dexamethasone on days 1-4, 9-12, and 17-20 of a 28-day cycle or on days 1, 8,15, and 22 of a 28-day cycle.
In another exemplary treatment regimen, the treatment regimen comprises administering 1.9mg/kg, 2.5mg/kg, or 3.4mg/kg of an anti-BCMA antigen binding protein on day 1 of a 28-day cycle; administering 10mg or 25mg of lenalidomide on days 1-21 of a 28 day cycle; and optionally, administering 20mg or 40mg of dexamethasone on days 1-4, 9-12, and 17-20 of a 28-day cycle or on days 1, 8,15, and 22 of a 28-day cycle.
Reagent kit
In some aspects, the present invention provides a kit for treating cancer, comprising:
(i) anti-BCMA antigen binding protein;
(ii) IMiD; and
(iii) instructions for treating cancer.
In some embodiments, the anti-BCMA antigen binding protein and IMiD are each formulated separately as their own pharmaceutical composition with one or more pharmaceutically acceptable carriers.
In some aspects, the present invention provides a kit for treating cancer, comprising:
(i) anti-BCMA antigen binding protein;
(ii)IMiD;
(iii) an anti-inflammatory compound; and
(iii) instructions for treating cancer.
In some embodiments, the anti-BCMA antigen binding protein, the ImiD, and the anti-inflammatory compound are each separately formulated with one or more pharmaceutically acceptable carriers as their own pharmaceutical composition.
In some aspects, the present invention provides a kit for treating cancer, comprising:
(i) anti-BCMA antigen binding protein;
(ii) instructions for treating cancer when combined with ImiD.
In some aspects, the present invention provides a kit for treating cancer, comprising:
(i) anti-BCMA antigen binding protein;
(ii) instructions for use in treating cancer when combined with an ImiD and an anti-inflammatory compound.
Examples
Example 1: multiple myeloma is treated with an anti-BCMA antibody drug conjugate, lenalidomide and dexamethasone.
A phase I/II study was conducted in human subjects to determine the safety, tolerability, and recommended phase 2 dose of anti-BCMA antigen binding protein in combination with lenalidomide and dexamethasone (RP2D) in relapsed/refractory multiple myeloma (RRMM) subjects, and to evaluate the safety and clinical activity of RP2D combination therapy in RRMM subjects.
The anti-BCMA antigen binding protein is an anti-BCMA antibody comprising a CDRH1 comprising the amino acid sequence set forth in SEQ ID No. 1; CDRH2 comprising the amino acid sequence set forth in SEQ ID NO. 2; CDRH3 comprising the amino acid sequence set forth in SEQ ID NO. 3; CDRL1 comprising the amino acid sequence set forth in SEQ ID NO. 4; CDRL2 comprising the amino acid sequence set forth in SEQ ID NO. 5; and CDRL3 comprising the amino acid sequence set forth in SEQ ID NO. 6; and conjugated to monomethyl auristatin f (mmaf), such as Tai et al blood.2014, 5 months and 15 days; 123(20): 3128 and 3138.
One treatment cycle was 28 days. Subjects who have not experienced dose limiting or intolerable adverse events can continue treatment with anti-BCMA antigen binding protein for up to 12 cycles and lenalidomide and dexamethasone for up to 14 cycles.
The study includes two parts: the first part is a dose escalation study and the second part is a dose extension study.
Study part 1 is a dose escalation phase used to assess safety and tolerability of the combined dose levels. It was aimed to identify the recommended phase 2 dose (RP2D) dose level of anti-BCMA antigen binding protein for use in combination with lenalidomide and dexamethasone. Initially the subject was on the first day of the 28 day cycle at 2.5mg/kg anti-BCMA antigen binding protein; on days 1 to 21 of a 28 day cycle with 25mg lenalidomide; and tested with 40mg dexamethasone on days 1, 8,15, and 22 of the 28 day cycle.
After cycle 1 is complete, the doses of anti-BCMA antigen binding protein, lenalidomide and dexamethasone drug can be adjusted as follows: the anti-BCMA antigen binding protein can be adjusted to 1.9mg/kg or 3.4 mg/kg; lenalidomide can be adjusted to 10 mg; and/or dexamethasone can be adjusted to 20 mg.
A summary of the treatment regimen is provided in table 1:
table 1: treatment regimens
Figure BDA0002412797080000301
In part 2 (dose extension), additional subjects were enrolled and treated with RP2D against each of BCMA antigen binding protein, lenalidomide and dexamethasone. Safety (AE, ECG, MM symptoms and laboratory assessments), clinical response and changes in symptoms/quality of life were assessed at the end of cycle 1 and all subsequent cycles.
Sequence listing
SEQ.ID.NO.1–CDRH1
NYWMH
SEQ.ID.NO.2:CDRH2
ATYRGHSDTYYNQKFKG
SEQ.ID.NO.3:CDRH3
GAIYDGYDVLDN
SEQ.ID.NO.4:CDRL1
SASQDISNYLN
SEQ.ID.NO.5:CDRL2
YTSNLHS
SEQ.ID.NO.6:CDRL3
QQYRKLPWT
Seq.id No. 7: heavy chain variable region
Figure BDA0002412797080000311
Sed.id.no. 8: light chain variable region
Figure BDA0002412797080000312
Seq.id No. 9: heavy chain region
Figure BDA0002412797080000313
Seq.id No. 10: light chain region
Figure BDA0002412797080000314

Claims (20)

1. A method of treating cancer in a subject in need thereof comprising administering a therapeutically effective dose of a combination comprising an anti-BCMA antigen binding protein and an immunomodulatory imide drug (IMiD).
2. The method of claim 1, wherein the combination further comprises an anti-inflammatory compound.
3. The method of claim 1 or claim 2, wherein the anti-BCMA antigen binding protein comprises a CDRH1 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in seq id No. 1; a CDRH2 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ id No. 2; 3 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ id No. 3; (ii) CDRL1 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ id No. 4; (ii) CDRL2 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ id No. 5; and a CDRL3 comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ id No. 6.
4. The method of any one of claims 1 to 3, wherein the anti-BCMA antigen binding protein is an antibody comprising a heavy chain variable region (VH) comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO 7; and a light chain variable region (VL) comprising an amino acid sequence having at least 90% sequence identity to the amino acid sequence set forth in SEQ ID NO. 8.
5. The method of any one of claims 2 to 4, wherein the anti-inflammatory compound is dexamethasone.
6. The method of any one of claim 1 to claim 5, wherein the IMiD is a thalidomide analog.
7. The method of any one of claims 1 to 5, wherein the IMiD is pomalidomide.
8. The method of any one of claims 1 to 5, wherein the IMiD is lenalidomide.
9. The method of any one of claims 1 to 8, wherein the anti-BCMA antigen binding protein is an immunoconjugate comprising an antibody conjugated to a cytotoxin.
10. The method of claim 9, wherein the cytotoxin is selected from MMAE or MMAF.
11. The method of any one of claims 1 to 10, wherein the cancer is selected from multiple myeloma, chronic lymphocytic leukemia, and non-hodgkin's lymphoma.
12. The method of any one of claim 1 to claim 11, wherein 1.9mg/kg, 2.5mg/kg, or 3.4mg/kg of anti-BCMA antigen binding protein is administered on day 1 of a 28-day cycle.
13. The method of any one of claim 1 to claim 12, wherein the IMiD is pomalidomide and wherein 4mg of pomalidomide is administered on days 1 to 21 of a 28-day cycle.
14. The method of any one of claim 1 to claim 12, wherein the IMiD is lenalidomide and wherein 10mg or 25mg of lenalidomide is administered on days 1-21 of a 28-day cycle.
15. The method of any one of claim 2 to claim 14, wherein the anti-inflammatory compound is dexamethasone and wherein 20mg or 40mg of dexamethasone is administered on days 1-4, 9-12, and 17-20 of a 28 day cycle or on days 1, 8,15, and 22 of a 28 day cycle.
16. A combination for use in the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, IMiD, and optionally, an anti-inflammatory compound.
17. Use of a combination for the manufacture of a medicament for the treatment of cancer, wherein the combination comprises an anti-BCMA antigen binding protein, an IMiD, and optionally an anti-inflammatory compound.
18. A kit for treating cancer, comprising:
(i) anti-BCMA antigen binding protein;
(ii) instructions for use in treating cancer when combined with an IMiD and optionally an anti-inflammatory compound.
19. A method of treating cancer in a human in need thereof comprising administering an anti-BCMA antibody drug conjugate and a thalidomide analog.
20. The method of claim 20, further comprising administering an anti-inflammatory compound.
CN201880060035.0A 2017-09-14 2018-09-12 Combination therapy for cancer Pending CN111107874A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762558593P 2017-09-14 2017-09-14
US62/558,593 2017-09-14
PCT/IB2018/056968 WO2019053612A1 (en) 2017-09-14 2018-09-12 Combination treatment for cancer

Publications (1)

Publication Number Publication Date
CN111107874A true CN111107874A (en) 2020-05-05

Family

ID=63713945

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201880060035.0A Pending CN111107874A (en) 2017-09-14 2018-09-12 Combination therapy for cancer

Country Status (7)

Country Link
US (2) US20200254093A1 (en)
EP (1) EP3691682A1 (en)
JP (2) JP2020533383A (en)
CN (1) CN111107874A (en)
BR (1) BR112020005077A2 (en)
CA (1) CA3075716A1 (en)
WO (1) WO2019053612A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114502593A (en) * 2019-08-06 2022-05-13 葛兰素史密斯克莱知识产权发展有限公司 Biopharmaceutical compositions and related methods
CA3151738A1 (en) * 2019-10-04 2021-04-08 Teru Hideshima Immunomodulatory imide drugs as zeta-chain-associated protein kinase 70 (zap70) agonists and uses thereof
CN114057890A (en) * 2020-07-31 2022-02-18 南京北恒生物科技有限公司 Novel costimulatory domains and uses thereof
CA3233953A1 (en) * 2021-10-05 2023-04-13 Matthew Bruce Combination therapies for treating cancer

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130280280A1 (en) * 2011-05-27 2013-10-24 Glaxosmithkline Antigen binding proteins
US20160297885A1 (en) * 2015-04-13 2016-10-13 Pfizer Inc. Therapeutic antibodies and their uses
GB201710620D0 (en) * 2017-07-03 2017-08-16 Glaxosmithkline Intellectual Property Ltd Targeted protein degradation

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
US6281230B1 (en) 1996-07-24 2001-08-28 Celgene Corporation Isoindolines, method of use, and pharmaceutical compositions
US5635517B1 (en) 1996-07-24 1999-06-29 Celgene Corp Method of reducing TNFalpha levels with amino substituted 2-(2,6-dioxopiperidin-3-YL)-1-oxo-and 1,3-dioxoisoindolines
DE69740095D1 (en) 1996-11-05 2011-02-17 Childrens Medical Center Thalidomide and dexamethasone for the treatment of tumors
US6045501A (en) 1998-08-28 2000-04-04 Celgene Corporation Methods for delivering a drug to a patient while preventing the exposure of a foetus or other contraindicated individual to the drug
US6020358A (en) 1998-10-30 2000-02-01 Celgene Corporation Substituted phenethylsulfones and method of reducing TNFα levels
US7629360B2 (en) 1999-05-07 2009-12-08 Celgene Corporation Methods for the treatment of cachexia and graft v. host disease
US6962940B2 (en) 2002-03-20 2005-11-08 Celgene Corporation (+)-2-[1-(3-Ethoxy-4-methoxyphenyl)-2-methylsulfonylethyl]-4-acetylaminoisoindoline-1,3-dione: methods of using and compositions thereof
US7893101B2 (en) 2002-03-20 2011-02-22 Celgene Corporation Solid forms comprising (+)-2-[1-(3-ethoxy-4-methoxyphenyl)-2-methylsulfonylethyl]-4-acetylaminoisoindoline-1,3-dione, compositions thereof, and uses thereof
US7968569B2 (en) 2002-05-17 2011-06-28 Celgene Corporation Methods for treatment of multiple myeloma using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione
US7230012B2 (en) 2002-11-14 2007-06-12 Celgene Corporation Pharmaceutical compositions and dosage forms of thalidomide
UA83504C2 (en) 2003-09-04 2008-07-25 Селджин Корпорейшн Polymorphic forms of 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione
TR201905423T4 (en) 2009-05-19 2019-05-21 Celgene Corp Formulations of 4-amino-2- (2,6-dioxopiperidin-3-yl) isindoline-1,3-dione
PT3415531T (en) * 2011-05-27 2023-09-12 Glaxo Group Ltd Bcma (cd269/tnfrsf17) - binding proteins
TWI679212B (en) 2011-11-15 2019-12-11 美商安進股份有限公司 Binding molecules for e3 of bcma and cd3
CA2869562C (en) 2012-04-11 2023-09-12 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Chimeric antigen receptors targeting b-cell maturation antigen
US10189906B2 (en) 2012-11-01 2019-01-29 Max-Delrück-Centrum Für Molekulare Medizin Antibody that binds CD269 (BCMA) suitable for use in the treatment of plasma cell diseases such as multiple myeloma and autoimmune diseases
TW201425336A (en) 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
US9243058B2 (en) 2012-12-07 2016-01-26 Amgen, Inc. BCMA antigen binding proteins
JP6636803B2 (en) 2013-02-05 2020-01-29 エンクマフ エスアーエールエル Method for selection of antibodies to BCMA
AR095374A1 (en) 2013-03-15 2015-10-14 Amgen Res (Munich) Gmbh UNION MOLECULES FOR BCMA AND CD3
GB201317928D0 (en) 2013-10-10 2013-11-27 Ucl Business Plc Molecule
AU2015248956B2 (en) 2014-04-14 2020-06-25 Cellectis BCMA (CD269) specific chimeric antigen receptors for cancer immunotherapy
JP6285274B2 (en) 2014-04-28 2018-02-28 株式会社ブリヂストン Bias tire and manufacturing method thereof
CN106687483B (en) 2014-07-21 2020-12-04 诺华股份有限公司 Treatment of cancer using humanized anti-BCMA chimeric antigen receptors
JP6706244B2 (en) 2014-07-24 2020-06-03 ブルーバード バイオ, インコーポレイテッド BCMA chimeric antigen receptor
EP2982692A1 (en) 2014-08-04 2016-02-10 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
EP3023437A1 (en) 2014-11-20 2016-05-25 EngMab AG Bispecific antibodies against CD3epsilon and BCMA
SI3226897T1 (en) 2014-12-05 2021-08-31 Memorial Sloan Kettering Cancer Center Antibodies targeting b-cell maturation antigen and methods of use
EP4310097A3 (en) 2014-12-05 2024-04-03 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting b-cell maturation antigen and uses thereof
SI3331910T1 (en) 2015-08-03 2020-07-31 Engmab Sarl Monoclonal antibodies against human b cell maturation antigen (bcma)
EP3147954A1 (en) 2015-09-22 2017-03-29 Nokia Technologies Oy Photodetector with conductive channel made from two dimensional material and its manufacturing method
CA3061945A1 (en) * 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130280280A1 (en) * 2011-05-27 2013-10-24 Glaxosmithkline Antigen binding proteins
US20160297885A1 (en) * 2015-04-13 2016-10-13 Pfizer Inc. Therapeutic antibodies and their uses
GB201710620D0 (en) * 2017-07-03 2017-08-16 Glaxosmithkline Intellectual Property Ltd Targeted protein degradation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
YU-TZU TAI等: "Targeting B-cell maturation antigen in multiple myeloma", 《IMMUNOTHERAPY》 *

Also Published As

Publication number Publication date
BR112020005077A2 (en) 2020-09-15
JP2023139179A (en) 2023-10-03
JP2020533383A (en) 2020-11-19
WO2019053612A1 (en) 2019-03-21
EP3691682A1 (en) 2020-08-12
US20200254093A1 (en) 2020-08-13
CA3075716A1 (en) 2019-03-21
US20240009307A1 (en) 2024-01-11

Similar Documents

Publication Publication Date Title
US11401334B2 (en) Combination treatment for cancer with anti-BCMA binding protein and proteosome inhibitor
US20240009307A1 (en) Combination Treatment for Cancer
US20220098302A1 (en) Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
CN109890387B (en) Use of glutamate modulators and immunotherapy for the treatment of cancer
JP2022515077A (en) CD70 and Venetoclax, BCL-2 Inhibitors, Combination Therapy for the Treatment of Acute Myeloid Leukemia
CN113924318A (en) Combination therapy with anti-BCMA antibodies and gamma secretase inhibitors
US20180235986A1 (en) Combination therapy for cancer treatment
AU2018266784A1 (en) LRRC33 inhibitors and use thereof
WO2018072743A1 (en) Use of pd-1 antibody conjugated with ido inhibitor in preparing anti-tumor drug
US20220411512A1 (en) Combination treatment for cancer
US20220003772A1 (en) Methods of treating cancer
KR20220004669A (en) Therapeutic Combinations Comprising Anti-CD123 Immunoconjugates
JPWO2020112796A5 (en)
CA3181535A1 (en) Methods for the use of a b7-h3 antibody-drug conjugate alone or in combination
NZ760009B2 (en) Means and methods for counteracting myeloproliferative or lymphoproliferative disorders

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination